Chemical Biology of N5

Chemical Biology of N5...
1 downloads 0 Views 2MB Size
Subscriber access provided by UB + Fachbibliothek Chemie | (FU-Bibliothekssystem)

Perspective 5

Chemical Biology of N-Substituted Formamidopyrimidine DNA Adducts Suresh S Pujari, and Natalia Y. Tretyakova Chem. Res. Toxicol., Just Accepted Manuscript • DOI: 10.1021/acs.chemrestox.6b00392 • Publication Date (Web): 21 Nov 2016 Downloaded from http://pubs.acs.org on November 28, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Chemical Research in Toxicology is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Chemical Biology of N5-Substituted Formamidopyrimidine DNA Adducts

Suresh S. Pujari and Natalia Tretyakova*

Department of Medicinal Chemistry and Masonic Cancer Center University of Minnesota, Minneapolis, Minnesota 55455, USA

*To whom correspondence should be addressed: Masonic Cancer Center, University of Minnesota, 2231 6th Street SE - 2-147 CCRB, Minneapolis, MN 55455, USA. phone: (612) 626-3432 fax: (612) 624-3869 e-mail: [email protected]

1 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 84

TOC Graphic:

2 ACS Paragon Plus Environment

Page 3 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Abstract: DNA nucleobases are the prime targets for chemical modifications by endogenous and exogenous electrophiles. Alkylation of the N7 position of guanine and adenine in DNA triggers base-catalyzed imidazole ring opening and the formation of N5-substituted formamidopyrimidine (N5-R-FAPy) lesions. Me-FAPy-dG adducts induced by exposure to methylating agents and AFB-FAPy-dG lesions formed by aflatoxin B1 have been shown to persist in cells and to contribute to toxicity and mutagenicity. In contrast, the biological outcomes of other N5substituted FAPy lesions have not been elucidated. To enable their structural and biological evaluation, N5-R-FAPy adducts must be site-specifically incorporated into synthetic DNA strands using phosphoramidite building blocks, which can be complicated by their unusual structural complexity. N5-R-FAPy exist as a mixture of rotamers and can undergo isomerization between α, β anomers and furanose-pyranose forms. In this Perspective, we will discuss the main types of N5-R-FAPy and summarize the strategies for their synthesis and structural elucidation. We will also summarize the chemical biology studies conducted with N5-R-FAPy-containing DNA to elucidate their effects on DNA and to identify the mechanisms of N5-R-FAPy repair.

3 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 84

List of Contents:

1.

Introduction

2.

Structural Identification and Synthesis of N5-Substituted FAPy Lesions (N5-R-FAPy) 2.1. Methyl-FAPy-dG 2.2. Ethyl-FAPy Adducts 2.3. 2-Hydroxyethyl-FAPy Adducts 2.4. 2-Oxoethyl-FAPy 2.5. Ethylamine-FAPy 2.6. FAPy Adducts of Nitrogen Mustards 2.7. Aflatoxin B1 (AFB 1)-FAPy-dG 2.8. Mitomycin C –FAPy Adducts

3.

Synthesis of DNA Oligodeoxynucleotides Containing N5-R-FAPy Adducts 3.1. Direct Alkylation of DNA to Generate N5-R-FAPy 3.2. Incorporation of Alkylated-FAPy Adducts into DNA via Phosphoramidite Chemistry 3.3. Carbocyclic Nucleoside Analogues of FAPy

4.

Effects of N5-R-FAPy Adducts on DNA Replication 4.1. Methyl-FAPy-dG 4.2. AFB-FAPy-G

5.

Cellular Repair of N5-R-FAPy Adducts

6.

Future Prospects and Outlook

4 ACS Paragon Plus Environment

Page 5 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

1.

Introduction N5-Alkyl-formamidopyrimidines (N5-R-FAPy) are ring open DNA adducts that form upon

imidazole ring opening of the corresponding N7-alkylpurine lesions.1-8 N7 positions of guanine and adenine in DNA are susceptible to electrophilic attack by a variety of alkylating agents. The resulting N7-substituted purines are destabilized due to the presence of positive charge at the N7 position9 and can undergo two competing reactions: depurination to form apurinic sites and imidazole ring opening to give N5-R-FAPy.1, 2, 10-13 While depurination is accelerated at low pH, N5-R-FAPy formation is preferred under basic conditions (Scheme 1). Although under physiological conditions, N5-R-FAPy adducts are formed in much lower yields than the corresponding depurinated adducts, they may have a significant biological impact because of their persistence in cells and their ability to induce mutations. Many simple alkylating agents including epoxides, nitrogen mustards, and alkyl halides preferentially alkylate the nucleophilic N7 position of guanine in DNA.14-21 However, not all of the resulting N7-dG adducts undergo imidazole ring opening under physiological conditions. Imidazole ring opening of N7-alkyl-dG is favored by electron withdrawing groups on the N7 substituent, which makes the C7-C8 bond more susceptible towards attack by hydroxyl anions.22, 23

Interestingly, imidazole ring opening of N7-alkyl-G adducts in RNA is 2-3 times faster than of

their DNA counterparts, presumably due to the electron withdrawing effect of the 2'-hydroxyl group.24 Aflatoxin B1 epoxide,25-27 N-methylnitrosamines,28-32 dimethyl sulfate,33, 34 tobacco carcinogen 4-(methylnitrosamino)-1-(pyridyl)-1-butanone (NNK),35 N-methylnitrosourea,36 1, 2dimethylhydrazine, N, N-dimethylnitrosamine,28-32, 37 cyclophosphamide,38, 39 mitomycin C,40, 41 and ethyleneimine42 are some examples of alkylating agents that give rise to N5 substituted FAPy adducts. N5-substituted FAPy adducts are also induced by leinamycin,20 pluramycins,43

5 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 84

azinomycin,44, 45 and S-(2-haloethyl)glutathione.46, 47 Structurally related unsubstituted FAPy adducts can be formed by a radical mechanism upon exposure to reactive oxygen species (ROS)48-50 but are beyond the scope of this review. Imidazole ring opening drastically changes the molecular shape and the hydrogen bonding characteristics of the parent purine nucleobase. Therefore, N5-substituted FAPy lesions are likely to induce DNA polymerase stalling, toxicity, and mutations. For example, N5-AFB1-FAPy adducts induced by Aflatoxin B1 are thought to play a major role in its hepatocarcinogenicity.51, 52

However, our understanding of the cellular formation and biological outcomes of N5-R-FAPy

adducts induced by other DNA modifying agents is incomplete. Chemical synthesis of N5-RFAPy nucleosides and N5-R-FAPy-containing DNA strands represents a special challenge due to the structural complexity of these unusual ring open lesions and their propensity to undergo isomerization. The goal of this Perspective is to summarize the current understanding of the mechanisms of formation, synthesis, isomerism, and biological consequences of N5-R-FAPy adducts. For additional information on N7-guanine alkylation and the chemistry of unsubstituted FAPy adducts induced by ROS, readers are referred to several recent reviews.7, 48-50, 53,54

2.

Structural Identification and Synthesis of N5-Substituted FAPy Lesions (N5-R-FAPy) Since their discovery in the early 1960s, N5-substituted FAPy adducts have been a subject

of intense investigation. In addition to simple DNA alkylating agents, a number of antitumor drugs and natural products have been shown to induce N5-R-FAPy adducts under physiological conditions, generating significant interest in these ring open structures. In this section, we will describe the discovery, mechanisms of formation, and synthesis of the major types of N5-RFAPy adducts.

6 ACS Paragon Plus Environment

Page 7 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

2.1. Methyl-FAPy-dG DNA can be methylated upon exposure to exogenous agents such as dimethyl sulfate,17, 33, 34

N-methylnitrosamines,28-32 and tobacco specific nitrosamine 4-(methyl- nitrosamino)-1-(3-

pyridyl)-1-butanone (NNK).35 DNA methylation can also occur from the reactions with endogenous enzyme cofactor, S-adenosyl-L-methionine (SAM).55 The most reactive site in DNA towards methylating agents is the N7 position of guanine.9, 16 Haines et al. described imidazole ring opening of N7-methylguanosine in the presence of ammonia or sodium hydroxide at room temperature.2 The ring open adduct was cleaved with acid, and the resulting aglycone was assigned the structure of 2,4-diamino-6-hydroxy-5N-methylformamidine (1 in Scheme 2) based on its spectroscopic properties.2 The biological significance of this finding remained unclear until 1983, when Beranek et al. isolated a novel nucleobase adduct from liver DNA of rats treated with the methylating agents, N, N,-dimethylnitrosoamine and 1,2-dimethyl hydrazine.37 The same adducts were subsequently found in bladder epithelial DNA of rats treated with N-methylnitrosourea.36 The unknown lesions were chromatographically identical to the ring opened derivative of 7methylguanine prepared by alkaline hydrolysis of N7-methyl-Guo, followed by cleavage of the glycosidic bond with acid (Scheme 2).36 Two isomers of the adduct were isolated by HPLC. Further experiments revealed that following isolation as two separate peaks, these two isomers interconverted to each other to give a 1:1 mixture.36, 37 Thermal desorption mass spectra of the two isomers were identical, giving a molecular ion peak m/z 183 and major fragments at m/z 155 and 140, corresponding to the loss of CO and CO+CH3, respectively.37 1H-NMR spectra of the two products were also identical, both exhibiting two distinct sets of resonances (Figure 1).36

7 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 84

NMR spectra of these isomers were consistent with cis and trans isomers around the C-N amide bond (Figure 2). NMR spectra revealed two sets of resonances each corresponding to two different forms of N5-methyl-N5-formyl-2,5,6-triamino-4-hydroxypyrimidine, which interconverted with each other.36 The relative abundances of the two rotamers were 1:9, 1:4, and 1:2 when spectra were taken in dimethylsulfoxide-d6, methanol-d4, and dimethylsulfoxided6/D2O, respectively (Figure 1).36 These results indicated that ring-open N5-methyl-N5-formyl2,5,6-triamino-4-hydroxypyrimidine adducts are found in vivo (and thus do not require strongly basic conditions to be formed) and exist as at least two interconverting forms (1 and 2 in Figure 2). The structural identities of the interconverting isomers of N5-methyl-FAPy adducts were further examined by proton NMR by Boiteux et al.10 Two methyl group signals were observed at 2.7 and 2.81 ppm, with the relative intensities of 88% and 12%, while the corresponding formamido signals were observed at 7.61 ppm (88%) and 7.88 ppm (12%) (Figure 3). Nuclear Overhauser Enhancement (NOE) was observed on the formamido proton at 7.61 ppm by irradiation of the methyl signal at 2.7 ppm, whereas no NOE effect was observed for formamido proton when methyl signal at 2.81 ppm was irradiated. This indicated that the protons observed as resonances at 2.7 ppm and 7.61 ppm are in a close proximity to each other and therefore belong to the Z conformer of the N-methyl-formamido bond, while the other isomers giving rise to resonances at 2.8 and 7.88 ppm is a E rotamer (3 and 4 in Figure 2). In summary, these early studies have revealed that, ring open N7-methyl-G adducts can form under physiological conditions. These adducts can exist as a mixture of four rotational isomers due to a hindered rotation about the C5-N5 and N-methyl-formamido bonds (1-4 in Figure 2). In the free nucleoside form these conformers rapidly interconvert with an estimated

8 ACS Paragon Plus Environment

Page 9 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

half-life of 8 min at 37 °C.10 Subsequently, these results were unambiguously supported by NMR studies of 15N-labeled adducts.56 As discussed below, the distribution of conformational isomers of N5-methyl-FAPy and other FAPy lesions is altered in double stranded DNA due to steric factors and hydrogen bonding interactions. The first synthesis of Me-FAPy-dG nucleoside was reported by Christov et. al. in 2008.57 In their approach, 2'-deoxyguanosine was protected at the exocyclic amine with N,Ndimethylformamide dimethylacetal and at the 5'-OH with DMT-Cl to give the doubly protected nucleoside 5 (Scheme 3). N7-methylation of 5 was carried out using CH3I in DMSO to give the N7-methyl-dG intermediate 6, which was not isolated (Scheme 3). Subsequent treatment of 6 with 1M NaOH and immediate neutralization yielded protected Me-FAPy-dG 7, which was characterized by NMR spectroscopy and high resolution mass spectrometry.57 The corresponding Me-FAPy-G ribonucleoside was synthesized using a similar route.58 N7-methylation of guanosine was achieved by reaction of unprotected guanosine with diazomethane, which was obtained from nitrosomethylurea as reported by Farmer et. al.59 Next, N7-methylguanosine was incubated with 0.15M ammonia for 30 min at 25 °C, yielding the Me-FAPy-G in 60% yield. The availability of Me-FAPy-dG and Me-FAPy-Guo has made it possible to incorporate these structures in nucleic acids chains using the phosphoramidite approach (Section 3 below).

2.2. Ethyl FAPy Adducts N7-ethylguanine is the main DNA lesion formed upon exposure to ethylating agents, and it can be converted to the corresponding Et-FAPy adducts.19, 58 van Delft et al. reported the synthesis of Ethyl-FAPy-dG (9 in Scheme 4) from N7-ethyl-dG 8 by basic treatment in 0.5 M ammonia for 70 min at 25 °C, followed by cooling to -80 °C and lyophilization (Scheme 4).58

9 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 84

The conversion yield was reported as 95%.58 The corresponding ribonucleoside (Et-FAPy-Guo) was prepared analogously by treating N7-Et-Guo with 0.1 N KOH at ambient temperature.19 A characteristic change in UV spectra was observed when N7-Et-Guo (λmax, 243 and 272 nm) was converted to Et-FAPy-Guo (λmax 265 nm in acid and 247 nm under basic conditions (pH 13, Figure 4).19

2.3. 2-Hydroxyethyl FAPy Adducts N7-(2-hydroxyethyl)guanine (N7-heG, structure 10a in Scheme 5) represents the major adduct produced upon exposure of DNA to ethylene oxide, which is commonly used as an intermediate in chemical industry. Ethylene oxide can also be formed endogenously through P450-mediated metabolism of ethylene.60, 61 As a result, N7-heG adducts are ubiquitously present in tissues of mice exposed to ethylene oxide and are among the most abundant endogenous DNA lesions measured, with > 30,000 N7-heG lesions per cell.53, 62 Roe et. al.63 reported the first synthesis of he-FAPy adducts. Guanosine was treated with ethylene oxide in glacial acetic acid at 50-55 °C to give N7-he-Guo (10b in Scheme 5).58 This intermediate was incubated with 0.5 M ammonia for 40 min at 25 °C, followed by lyophilization to give N7-(2-hydroxyethyl)-FAPy-Guo (compound 11b).63 The 2’-deoxy counterparts 10a and 11a were prepared in a similar fashion.63

2.4. 2-Oxoethyl-FAPy Vinyl chloride is an important industrial chemical classified as a known carcinogen.64-67 It is epoxidized by cytochrome P450 2E1 to chlorooxirane, which reacts extensively with DNA.68 Acetoxyoxirane, an acetylated analogue of chlorooxirane, was used in many studies of DNA

10 ACS Paragon Plus Environment

Page 11 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

alkylation because it is more chemically stable than chlorooxirane, but generates the same types of DNA adducts.69 Acetoxyoxirane can be readily prepared from vinyl acetate and dimethyldioxirane.69 Similar to other simple epoxides, chlorooxirane preferentially alkylates N7G in DNA, and the resulting adducts can undergo imidazole ring opening.69 Christov et. al. reported the first synthesis of oxoethyl-FAPy-dG (Scheme 6).69 In brief, dG was reacted with acetoxyoxirane in acetic acid at room temperature for 3 h to give N7-oxyethyl-dG 12. At pH 7.8, compound 12 was found to undergo two competing reactions: deglycosylation to give an abasic site (major pathway) and imidazole ring opening to give compound 13 in 10 % yield (Scheme 6, top). Compound 13, which was accounted for 10% of total reaction mixture, was highly unstable and underwent spontaneous cyclization to give a ring closed compound (cyclized product, Scheme 6). In a similar way the allyl-FAPy-dG was obtained by the alkylation of dG with allyl bromide to give compound 14 which was further treated with 1M NaOH to give the corresponding FAPy adduct 15 (Scheme 6, bottom).

2.5. Ethylamine-FAPy Ethyleneimine (aziridine) is an industrial chemical widely used in the production of polymers, coatings, adhesives, drugs, dyes, cosmetics, and antineoplastic agents.70, 71 Carboxylic acid derivatives of aziridine have been reported as immunomodulators.72 Ethyleneimine is an extremely reactive alkylating agent which targets the N7- purine positions in DNA. In 1984, Hemminki et. al.42 reported that ethyleneimine reactions with guanosine and 2'-deoxyguanosine at pH 6.5 in 0.2 M ammonium formate for 6 h gave rise to N7-dG adducts 16a or 16b (Scheme 7).42 The corresponding ring opened adducts 17a or 17b were formed in 80% yield. Unlike N7

11 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 84

adducts of simple alkylating agents, the conversion of aziridine adducts to the corresponding FAPy structures took place under mild conditions, probably due to protonation of the amino side chain, which facilitates hydroxyl ion attack at C-8 position of N7-alkylated nucleoside (Scheme 7). As described below, a similar mechanism accelerates FAPy adduct formation from antitumor nitrogen mustards.

2.6. FAPy Adducts of Nitrogen Mustards Nitrogen mustards (NM) are bis-electrophiles capable of cross-linking DNA to form toxic interstrand and intrastrand cross-links (Figure 5). Nitrogen mustard drugs such as cyclophosphamide, chlorambucil, and mechlorethamine (18-21) are widely used in the treatment of immune diseases, lymphoma, leukemia, multiple myeloma, and ovarian carcinoma (Figure 5).73-76 The antitumor activity of NMs has been attributed to their ability to cross-link the twin strands of DNA.77 The resulting bifunctional lesions, if not repaired, can inhibit DNA replication and transcription, eventually leading to cell cycle arrest, apoptosis, and the inhibition of tumor growth. In 1982, Chetsanga et. al. first reported imidazole ring opening of N7-guanine adducts generated by phosphoramide mustard in DNA.78 Alkylated DNA was treated with 0.2 N NaOH for 30 min at 37 °C to obtain the corresponding FAPy adducts. The reaction mixture contained five isomers of phosphoramide mustard-imidazole ring-opened dG complexes.78 Recently, the Turesky group for the first time detected NM-FAPy adduct in mustard-treated DNA and in human cell culture.79 Christov et al. recently reported the first synthesis of NM-FAPy adducts, which were subsequently incorporated into DNA strands by phosphoramidite chemistry (Section 3 below).80 The synthesis began with N2 – formamidine protected compound 22, which was further protected

12 ACS Paragon Plus Environment

Page 13 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

at 5’OH to give DMT protected dG (23, Scheme 8). Compound 23 was reacted with ethyl nitrogen mustard in trifluoroethanol to give N7-dG intermediate 24, which was not isolated. Further imidazole ring opening of 24 was performed in the presence of 1M NaOH to give NMFAPy-dG 25 (85% yield).80

2.7. Aflatoxin B1 (AFB 1)-FAPy-dG Aflatoxins are carcinogenic mycotoxins produced by certain molds that can contaminate agricultural products such as peanuts and corn.81, 82 Specifically, Aflatoxin B1 is produced by Aspergillus flavus and A. parasiticus and has been implicated in liver cancer in populations consuming contaminated grains.83 Aflatoxin B1 is metabolically activated to epoxide 26 (Figure 6), which is capable of alkylating guanine nucleobases of DNA to give N7-guanine adducts 27, which spontaneously depurinate to give abasic sites.84 Aflatoxin exposure induces high levels of G→T transversions.85-88 AFB-N7-dG adducts were initially hypothesized to be responsible for these genetic changes.83, 89, 90 However, sitespecific mutagenesis experiments have revealed that N7-dG adducts 27 are only weakly mutagenic in E. coli. (4%).91 It was then proposed that apurinic (AP) sites arising from depurination of N7-guanine adducts may be the source for AFB genotoxicity.14, 92 However, Essigmann and coworkers have shown that AFB 1 epoxide treated cells exhibit a unique mutagenic signature distinct from that of AP sites.91 It was subsequently shown that N7-AFB-dG undergoes imidazole ring opening at physiological pH to give the corresponding AFB-FAPy-dG adduct 28.89, 90 AFB-FAPy-dG formation occurs readily at physiological pH,83 and the resulting lesions are highly mutagenic84, 90, 93-95 and persist in vivo.84, 93, 94 It is now generally accepted that

13 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 84

ring open adducts AFB-FAPy-dG (28 in Figure 6) are largely responsible for the key G to T mutations that lead to hepatocarcinoma development following exposure to Aflatoxin B1.96 The first synthesis of AFB-FAPy-dG adducts was reported by Harris and coworkers (Scheme 9).82 5'-DMT-protected-2'-deoxyguanosine was treated with AFB epoxide 26 in THF to give the corresponding N7-dG adduct 29. Protected AFB-FAPy-dG 30 was obtained by incubating compound 29 at pH 9.5 overnight (15 mM Na2CO3/30 mM NaHCO3 buffer at ambient temperature). Subsequent detritylation of compound 30 in 0.1 M HCl for 15 min gave AFB-FAPy-dG nucleoside 31.82 The use of 5'-protected dG was critical to prevent anomerization of the sugar (see below Section 3).82 The presence of multiple isomeric forms of AFB-FAPy-dG has made it challenging to deduce the exact chemical structure of AFB-FAPy-dG. Initial NMR studies by Miller and Garner groups have revealed that AFB-FAPy-dG existed as a mixture of at least four isomers.89, 90 These four isomers were isolated by HPLC as two separate peaks, with each fraction containing a pair of inseparable species. Miller and Garner initially hypothesized that the structures of the AFBFAPy isomers involved ring closed forms 32B and 33B, along with the corresponding ring open structures 32A and 33A (Figure 7).89, 90 Subsequent structural studies by Hertzog et. al.90 have reassigned the structures of AFB-FAPy isomers as two pairs of geometrical isomers 34 and 35, which were in equilibrium with the rotamers 36 and 37 (Figure 8). UV absorption spectra of 34 and 35 were reported to have maxima at 265, 340 and 364 nm, with pH having only a minor effect on the spectra.71 However, the presence of structural isomers 34 and 35, with different position of the formyl group (N5 vs N6), due to different direction of imidazole ring opening following hydroxyl ion attack, was in contrast with previous studies of FAPy adducts generated

14 ACS Paragon Plus Environment

Page 15 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

by methylating agents, sterigmatocystin, and mitomycin, in which the formyl group was always placed at the N5 atom as in 34 and 36.10, 36, 41, 97 Most recently, detailed NMR experiments were conducted by the Harris group82 to elucidate the correct structure of AFB-FAPy adducts. (in blue in Figure 9). Two sets of stereoisomers are available for AFB-FAPy adducts; (a) geometrical isomers around formamide group and, (b) atropisomers arise due to hindered rotation around the pyrimidine C5-N5 bond. The two pairs of AFB-FAPy isomers which form as a result of hindered rotation about the C5-N5 bond (38 and 39; in blue, Figure 9) are atropisomers.82 The steric bulk of the AFB substituent at N5 creates a high rotational barrier for this rotation. Since aflatoxin is a chiral molecule, this leads to a pair of diastereomers separable by HPLC. These two pairs of compounds appeared as four species with their respective isomers 40 and 41, which form as a result of rotation about the N-methyl-formamido bond (highlighted in red in Figure 9). 1H-NMR spectra of stereoisomers 38 and 39 are very similar. A pair of formyl signals are observed at 8.29, 7.59 ppm (compound 38) and 8.22, 7.52 ppm (compound 39). These four formyl signals are split into doublets with coupling constants ~17 Hz, confirming the attachment of formyl group to the N5. Similar observations were made for FAPy nucleoside 31 (AFB-FAPy-dG). The complete NMR studies of AFB-FAPy-dG included COSY, TOCSY, HMQC, HMBC, NOESY and ROESY.82 Structurally related FAPy adducts of sterigmatocystin, a mycotoxin that can form in moldy grain, green coffee, and cheese, have been reported by Baertschi et. al.98 Sterigmatocystin is produced by some strains of Aspergillus, Penicillium, and Bipolaris sp.99, 100 Sterigmatocystin 1,2 epoxide 42 (Scheme 10) was prepared from sterigmatocystin and incubated with DNA for 7 days at 5 °C to form the corresponding alkylated adduct 43. Ring opening of this adduct was

15 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 84

performed by incubating compound 43 in pH 9.8 at 25 C for 2 hr to obtain sterigmatocystinFAPy 44 (Scheme 10).97, 99, 100

2.8. Mitomycin C-FAPy Adducts Mitomycin C (MMC, in Scheme 11) is an antitumor antibiotic widely used in cancer chemotherapy.101 It is a bifunctional alkylating agent capable of cross-linking DNA, leading to cell cycle arrest and apoptosis. MMC alkylates N7-G position in DNA to give the corresponding alkylated adduct.40 In 1987, Tomasz et. al. reported that the mitomycin C forms FAPy-dG adducts under basic conditions.41 d(GpC) dinucleotide was treated with MMC at pH 3.5-4 to give the corresponding N7-alkylguanine adduct 45. Subsequent imidazole ring opening under basic condition yielded MMC-FAPy-dG 46 (Scheme 11). Five isomers of MMC-FAPy-dG (47-50) were identified, including α and β anomers and furanose/ pyranose nucleosides (Figure 10).41 The α anomer 53 may form through imino intermediate 52 (Scheme 12), a rearrangement that can take place in double stranded DNA, but is much faster for free nucleosides. The pyranose isomers of FAPy-dG adducts (54 in Scheme 12) may form by intramolecular attack of C5'-hydroxyl on the imine intermediate 52.41, 102 It should be noted that the rearrangement to pyranose form is not possible in the absence of a free C5'hydroxyl group (as in DNA strands).7, 103 Therefore, during nucleoside and phosphoramidite synthesis of FAPy adducts, the formation of pyranose isomers can be minimized by protecting the 5'position of the sugar, typically using the DMT group.57

3.

Synthesis of DNA Oligodeoxynucleotides Containing N5-R-FAPy Adducts

16 ACS Paragon Plus Environment

Page 17 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

In order to establish the role of N5-substituted FAPy adducts in mutagenicity and to uncover their possible contributions to the therapeutic effects of DNA modifying agents, it is necessary to establish the chemical structures, stability, and mispairing characteristics of N5-RFAPy adducts. This necessitates chemical synthesis of DNA molecules containing structurally defined, site-specific N5-R-FAPy adducts. Such a synthesis can be a challenging task due to the unusual structural complexity of this class of adducts and their ability to undergo isomerization.82 Standard solid phase synthesis coupling conditions can result in a mixture of DNA strands containing α and β anomers, as well as both furanose and pyranose forms, and special precautions must be taken to minimize this structural complexity. Previous attempts to prepare N5-R-FAPy containing DNA strands can be broadly divided into three general approaches; (i) direct treatment of DNA with alkylating agents and base to introduce FAPy, (ii) chemical synthesis of alkylated-FAPy-phosphoramidite building blocks and their incorporation into DNA via solid phase synthesis (SPS), and (iii) solid phase synthesis employing carbocyclic nucleoside analogues.

3.1. Direct Alkylation of DNA to Generate N5-R-FAPy In the most straightforward approach, DNA strands containing a FAPy adduct can be prepared by treating oligodeoxynucleotides containing a single guanine base with alkylating agents, followed by basic treatment to introduce N5-R-FAPy (Scheme 13). This methodology was employed by Brown et al. to generate DNA containing AFB-FAPy-dG.82 Synthetic DNA 13-mer containing a single dG residue was treated with AFB-epoxide in 100 µL phosphate buffer (10 mM sodium phosphate, 100 mM NaCl, pH 7.0) for 30 min (Scheme 14). Further, the alkylated DNA was dissolved in sodium carbonate solution (pH 10) to open the imidazole ring,

17 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 84

and the resulting AFB-FAPy-dG containing oligodeoxynucleotide was purified by HPLC. The main limitations of this approach are that it is limited to DNA sequences containing a single guanine and that a mixture of isomers can be generated. Tudek et al.104 employed a similar direct alkylation approach to study the mutagenic specificity of Me-FAPy-purines in M13mp18 phage DNA. Single stranded M13 phage DNA was incubated with dimethylsulfate (DMS) to obtain DNA containing N7-methyl-dG (83%), and N7-methyl-dA (2.2%) (Scheme 15). This DNA was further incubated in 0.2M NaOH for 15 min at 37 °C to obtain DNA containing the corresponding FAPy adducts.104 It should be noted that this approach produced Methyl-FAPy-dG and Methyl-FAPy-dA adducts at many different sites within the plasmid. Chetsanga et. al. treated DNA containing [3H]-dG with phosphoramide mustard. To obtain labelled DNA, a guanine requiring Bacilius Subtilis strain was grown in cell culture supplemented with deoxy[8-3H]guanosine, and cells were lysed by lysosome treatment.78 The purified DNA consisting [3H]guanosine was treated with phosphoramide mustard to obtain alkylated DNA and with 0.2N NaOH to produce PM-FAPy containing DNA (Scheme 16). As in the paper by Tudek et al., this approach does not produce site-specific adducts.104

3.2. Incorporation of Alkylated-FAPy Adducts into DNA via Phosphoramidite Chemistry To enable the preparation of DNA strands containing site specific, structurally defined N5R-FAPy adducts, solid phase synthesis starting with nucleoside phosphoramidites can be employed. In 2008, the Rizzo group reported the synthesis of Me-FAPy-dG phosphoramidite (Scheme 17). Compound 55 was further treated with phosphoramidite reagent in the presence of

18 ACS Paragon Plus Environment

Page 19 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

tetrazole in anhydrous dichloromethane for 2 h at room temperature to obtain Me-FAPy-dG phosphoramidite 56 in 78% yield (Scheme 17).57 This phosphoramidite building block was employed in solid phase synthesis (SPS) experiments in order to incorporate Me-FAPy-dG into short DNA sequences to generate the 5'-d(TT-Me-FAPy-dG-TTC)-3' modified oligodeoxynucleotides. The critical step in the solid phase synthesis of Me-FAPy-dG containing ODNs is the deprotection of the 5'-OH group of Me-FAPy-dG, since the unprotected nucleoside undergoes rapid rearrangement to the pyranose form (Scheme 12 and discussion above).57 During solid phase synthesis, a “short” deprotection step was employed rather than the traditional deprotection step.57 Upon HPLC analysis of enzymatic digests, five Me-FAPy-dG peaks were observed (Figure 11), of which one peak was the pyranose form (minor amount) whereas other four peaks were furanose nucleosides in an α and β anomeric forms.57 A similar strategy was employed to prepare NM-FAPy-dG building block (59 in Scheme 18).80 The protected NM-FAPy-dG (compound 25) was prepared as shown in Scheme 8. Compound 25 was then treated with cesium acetate in 18-crown-6 ether to replace chloro group of NM with an acetyl group (57). The 6-oxo group was deprotected with TBAF in THF to give compound 58, which upon phosphitylation in dichloromethane in the presence of tetrazole gave phosphoramidite building block 59 in 50% yield. This nucleoside was incorporated into 12 and 24-mer oligodeoxynucleotides by solid phase synthesis.80 During automated solid-phase synthesis, a short deprotection cycle was employed in order to minimize the rearrangement of NM-FAPy-dG to the pyranose form of the nucleoside. However, HPLC analysis of the resulting DNA strands revealed two peaks, both having the expected molecular weight and representing both furanose and pyranose forms. When the

19 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 84

standard DNA synthesis protocol (normal deprotection time) was employed, a 1:1 ratio of furanose to pyranose ring containing product formation was observed. The thermal melting profile of the NM-FAPy-containing 12 mers gave inconsistent results due to the presence of α and β anomers (1:1).80

3.3. Carbocyclic Nucleoside Analogues of FAPy As described above, a major obstacle in solid phase synthesis of FAPy-dG containing DNA strands is that they readily undergo rapid isomerization to give α anomers and pyranose forms under standard DNA synthesis conditions.105, 106 Due to this rapid anomerization, it has only been possible to incorporate α/β anomeric mixtures of FAPy adducts into DNA. Hydrolysis of glycosidic bond of FAPy-dG at elevated temperatures further complicates their synthesis. To circumvent these problems, Carell et al. have developed the nonhydrolizable and nonepimerizable β analogues of the FAPy-dG lesions.105-107 In this approach, the oxygen atom of the deoxyribose moiety was replaced with a methylene group to give the corresponding carbocyclic nucleoside. This replacement has a minor effect on base pairing.59 The synthesis of Bz-cFAPy-dG (Scheme 19)105-107 started with enantiomerically pure cyclopentylamine 60, which was synthesized as described by Cullis and Dominguez.108 Coupling of compound 60 with protected 2-amino-6-chloro-5-nitro-4-oxopyrimidine (61) furnished the nitro pyrimidine derivative 62 (86%), which was subsequently subjected to reduction to give the corresponding aminopyrimidine compound 63 (58%). Further, the primary amine of compound 63 was coupled with benzaldehyde and subjected to reduction with sodium cyanoborohydride to give the benzylated compound 64. The formyl group was introduced at the C-5 position by reaction of compound 64 with mixed anhydride to give Bz-cFAPy-dG 65 (89%) (Scheme 19).

20 ACS Paragon Plus Environment

Page 21 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

To prepare the nucleoside phosphoramidite, compound 65 was protected at 5'-OH by DMTCl in pyridine at room temperature to give the DMT derivative 66 in 55% yield (Scheme 19). Phosphitylation of 66 was carried out under argon in acetonitrile in the presence of tetrazole and DIPEA to give phosphoramidite 67 in 54% yield, which was subsequently incorporated into DNA. These authors reported that since the standard capping procedure during DNA synthesis was not compatible with Bz-cFAPy-dG, a solution of 2,6-lutidine and pivaloylic anhydride in THF (v/v/v 1:1:8) was used in place of phenoxyacetyl anhydride for capping, and 4,5dicyanoimidazole (0.25 M) was used as the coupling reagent. The coupling time for the BzcFAPy-dG phosphoramidite was much longer compared to standard phosphoramidite methods (10 min vs 144 s).106 DNA strands containing Bz-cFAPy-dG were purified by HPLC and analyzed by MALDITOF. LC-MS of enzymatic digests showed no structural alteration of Bz-FAPy-dG during ODN synthesis and purification. Thermodynamic studies of c-FAPy (no substitution on the formamide group) oligodeoxynucleotides revealed that c-FAPy conferred significant duplex destabilization.105, 106 Interestingly, the base excision repair enzyme Fpg recognized the unnatural N7-benzyl-cFAPy-dG lesion via an unproductive binding mode, leading to enzyme inhibition.107

4.

Effects of N5-R-FAPy Adducts on DNA Replication 4.1. Methyl-FAPy-dG Structurally, ring open N5-R-FAPy adducts are substituted pyrimidines, and are expected to

mispair with purines during DNA replication. However, initial studies with bacterial DNA polymerases (e.g. Klenow fragment of E. coli. DNA polymerase I) have shown that MethylFAPy-dG blocked bacterial DNA replication in vitro, but did not induce any mutations.11, 53, 109,

21 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

110

Page 22 of 84

Similarly, O'Connor and others reported that E. coli. DNA polymerase I exo and T4 DNA

polymerases were inhibited one nucleotide before Me-FAPy-G.11 Inhibition of DNA synthesis by Me-FAPy-G is stronger than ROS-induced lesion 8-oxo-dG, but is weaker than that of apurinic sites and FAPy-A (FAPy-Ade ≈AP site>FAPy-7Me-G>8-oxoG).54, 111 Rizzo et al. examined in vitro bypass of Me-FAPy-dG in the presence of eukaryotic DNA polymerases α, β, and hPol δ/PCNA.112 Me-Fapy-dG blocked high fidelity polymerases at either the insertion or the extension step. Translesion synthesis was observed for hPols η, κ, and hRev1/Pol ζ. These polymerases primarily inserted the correct base (C) opposite the lesion, however hPols η and κ also catalyzed the misinsertion of Thy, Gua, and Ade opposite Me-FapydG, and generated a single nucleotide deletion product. These authors concluded that although the amounts of Me-FAPy-dG lesions in cells are low, their miscoding potential could contribute to genomic instability.112 Tudek et. al.111, 113 investigated the biological properties of Me-Fapy-dG in M13mp18 phage DNA. Lesions containing plasmids were generated as described above in section 3.1 (Scheme 15), and are transfected into E. coli. The presence of Me-FAPy adducts led to a significant decrease in transfection efficiency and increased mutational frequency in the lacZ gene following SOS induction.111, 113 However, sequencing analyses have revealed primarily A →G transitions, while mutations at GC base pairs were only slightly elevated. These results suggest that Me-FAPy-G is primarily a lethal lesion in E. coli. In contrast, the corresponding MeFAPy-A is a more miscoding lesion, causing the A→G transitions.54, 104 Me-FAPy-A (Figure 12) was two orders of magnitude more mutagenic than Me-FAPy-G.54, 111

4.2. AFB-FAPy-G

22 ACS Paragon Plus Environment

Page 23 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

The biological outcomes of AFB1-FAPy-dG adducts (Figure 13) have been examined in detail due to their proposed roles in aflatoxin-mediated liver cancer. In contrast to N7-AFB-G, AFB1-FAPy-G is highly persistent in rat liver DNA, reaching maximum adduct amounts 2 weeks after exposure.89 One of the AFB1-FAPy rotamers (68A in Figure 13) has been shown to be a potent block to DNA synthesis, even when DNA polymerase of lowered replication fidelity was used (MucAB).96 Both AFB1-FAPy-G and N7-AFB1-G cause G→T transversions, which is consistent with the observed G→ T mutations in codon 249 of the p53 tumor suppressor gene in 50% of hepatocellular carcinomas and in AFB1-treated human hepatocytes cultures.51, 52 In addition, AFB1-induced G→ T mutations in the ras oncogene appear to be important for liver tumor progression.86, 87 Taken together, these results indicate that AFB1-FAPy adducts may be the ultimate lesions responsible for mutagenesis and genotoxicity of aflatoxin.96, 114

5.

Cellular Repair of N5-R-FAPy Adducts Many of the previous studies of FAPy adduct repair have been limited to unsubstituted

FAPy induced by ROS.49, 50, 53 Repair studies of N5-substituted FAPy-adducts are less extensive, and several examples are given below. FPG glycosylase: Formamidopyrimidine DNA glycosylase (Fpg) was first identified in 1978-1979 as a DNA glycosylase that removes Me-FAPy-G from DNA.13 Along with MeFAPy-G, this glycosylase also excises ROS-induced unsubstituted FAPy-G, unsubstituted FAPyA, as well as damaged pyrimidines and 8-oxo-dG.13,115-119 Substituent size on the N5 position of FAPy has been shown to influence enzyme activity. For example, Tudek et. al. have shown that Me-FAPy-G was excised by Fpg 7-times faster than Et-FAPy-G.120 It is not known whether other N5-R-FAPY adducts are also substrates for this repair pathway.

23 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 84

yOgg 1: 8-oxo-G DNA glycosylase (yOgg 1) excises Me-FAPy-G, but does not remove MeFAPy-A.121, 122 Furthermore, Fpg and its eukaryotic homolog Ogg1 have been reported to recognize unsubstituted FAPy-dG and the carbocyclic analog of Bz-FAPy-dG (Bz-cFAPy-dG, Figure 14 selectively recognizes rotamer 1) with high affinity. hNEIL and mNEIL 1: Both hNEIL 1 (human NEIL 1) and mNEIL 1 (mouse NEIL 1) excised Me-FAPy-G, along with a number of pyrimidine-derived nucleobase lesions. However, hNEIL is the only human enzyme that excises FAPy-Ade (unsubstituted).123-127 E. coli. Endonuclease IV: E. coli. Endonucleoase IV (Endo IV) is an AP endonuclease specific for double stranded DNA . It can also remove the 3'-blocking phosphate groups128-130 and possess the 3' →5' endonuclease activity.131, 132 ODNs containing α-dA, α-dT, α-dC, α-FAPy-dG, αFAPy-dA lesions are substrates to Endo IV.133-137 Asagoshi et. al. reported that oligodeoxynucleotides containing α-Me-FAPy-dG are not substrates for Endo IV.110, 138 However, in 2000 Christov and others showed that α-Me-FAPy-dG is indeed a substrate for Endo IV. 139 The Me-FAPy-dG lesion is a substrate for Fpg/Nei family of glycosylases as well. It is possible that Endo IV and Fpg glycosylases play specialized roles in FAPy adduct repair, with Endo IV recognizing only the α-anomer of lkayl-FAPy lesions and Fpg glycosylase recognizing the β-anomer form of the adduct. Nucleotide excision repair Alekseyev and Essigmann reported that Aflatoxin B1 formamidopyrimidine adducts (AFB1-FAPy-dG, Figure 13) are preferentially repaired by the nucleotide excision repair pathway in vivo.140 These authors transfected plasmids containing sitespecific AFB1-FAPy-dG lesions into E.coli cells and employed the host cell reactivation assay to monitor lesion repair in wild type cells and their repair deficient clones. Cells deficient in nucleotide excision repair (uvrA) were unable to remove the damage, while BER mutants (mutM)

24 ACS Paragon Plus Environment

Page 25 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

were affected to a lesser extent.140 This was confirmed by in vitro experiments with sitespecifically modified oligodeoxynucleotides and purified MutM protein, which revealed excision products characteristic of NER.140

6.

Future Prospects and Outlook Although the chemistry and biology of the N5-substituted FAPy lesions have been a

subject of interest for several decades, the majority of the published studies have focused on two types of FAPy lesions (Me-FAPy and AFB-FAPy). The chemical biology of other N5-R-FAPy adducts is incompletely understood, and their roles in chemical carcinogenesis remain unknown. Based on the published studies of Me-FAPy and AFB-FAPy, such lesions may be extremely important for the biological mechanisms of many DNA damaging agents, if formed in vivo. However, with a few exceptions, it is not known whether significant amounts of N5-R-FAPy adducts form in cells treated with DNA alkylating drugs and environmental agents. Future mass spectrometry based studies are urgently needed to establish the concentrations of these secondary adducts in cells and tissues. Chemical synthesis of DNA strands containing structurally defined N5-R-FAPy represents a significant challenge due to their propensity to undergo isomerization. Furthermore, FAPy lesions exist as a mixture of rotamers which present a range of possibilities for base pairing due to changes in hydrogen bond donor acceptor patterns. Future studies employing novel solid phase synthesis methodologies are needed in order to establish the relationship between N5-R-FAPy adduct structures and their biological outcomes, as well as to elucidate their effects on DNA structure.

25 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 84

Funding information: Funding for this research was provided by grants from the National Cancer Institute (CA1006700), the National Institute of Environmental Health Sciences (ES023350), and University of Minnesota College of Pharmacy.

Acknowledgements: We thank Arnold Groehler IV (University of Minnesota) for helpful discussions and Bob Carlson (Masonic Cancer Center, University of Minnesota) for preparing figures for this manuscript and editorial assistance.

Abbreviations: FAPy, formamidopyridines; dG, 2'-deoxyguanosine; dA, 2'-deoxyadenosine; DNA, deoxynucleic acid; N7-heG, N7-(2-hydroxy)ethyl guanine; PM, phosphoramide; NM, nitrogen mustard; MMC, mitomycin C; AFB, aflatoxin; THF, tetrahydofuran; DCM, dichloromethane; CH3I, methyliodide; HCl, hydrochloric acid; DMSO, dimethylsulfoxide; NaOH, sodium hydroxide; AcOH, acetic acid; EI, ethyleneimine; DMTCl, dimethoxytrityl chloride; HPLC, high performance liquid chromatography; UV, ultraviolet; NMR, nuclear magnetic resonance; ppm, parts per million; Me, methyl; ROS, reactive oxygen species; ODN, oligodeoxynucleotides; DMS, dimethylsulfate; Na2CO3, sodium carbonate; NaHCO3, sodium bicarbonate; AFB-FAPy, aflatoxin formamidopyrimidine; Me-FAPy, methyl formamidopyrimidine, Et-FAPy, ethyl formamidopyrimidine; MMC-FAPy, mitomycin C formamidopyridine; NM-FAPy, nitrogen mustard formamidopyridine; PM-FAPy, phosphoramide formamidopyridine; Bz-cFAPy; benzyl carbocyclic formamidopyridine

26 ACS Paragon Plus Environment

Page 27 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

References: (1)

Townsend, L. B., and Robins, R. K. (1963) Ring Cleavage of Purine Nucleosides to Yield Possible Biogenetic Precursors of Pteridines and Riboflavin. J. Am. Chem. Soc. 85, 242-243.

(2)

Haines, J. A., Reese, C. B., and Todd, L. (1962) 1009. The methylation of guanosine and related compounds with diazomethane. J. Chem. Soc. (Resumed), 5281-5288.

(3)

Chetsanga, C. J., Bearie, B., and Makaroff, C. (1982) Alkaline opening of imidazole ring of 7-methylguanosine. 1. Analysis of the resulting pyrimidine derivatives. Chem. Biol. Interact. 41, 217-233.

(4)

Chetsanga, C. J., and Makaroff, C. (1982) Alkaline opening of imidazole ring of 7methylguanosine. 2. Further studies on reaction mechanisms and products. Chem. Biol. Interact. 41, 235-249.

(5)

Hecht, S. M., Adams, B. L., and Kozarich, J. W. (1976) Chemical transformations of 7,9disubstituted purines and related heterocycles. Selective reduction of imines and immonium salts. J. Org. Chem. 41, 2303-2311.

(6)

Darzynkiewicz, E., Labadi, I., Haber, D., Burger, K., and Lonnberg, H. (1988) 7Methylguanine nucleotides and their structural analogues; protolytic equilibria, complexing with magnesium (II) ion and kinetics for alkaline opening of the imidazole ring. Acta Chem. Scand. B. 42, 86-92.

(7)

Gates, K. S., Nooner, T., and Dutta, S. (2004) Biologically relevant chemical reactions of N7-alkylguanine residues in DNA. Chem. Res. Toxicol. 17, 839-856.

27 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(8)

Page 28 of 84

Gates, K. S. (2009) An overview of chemical processes that damage cellular DNA: spontaneous hydrolysis, alkylation, and reactions with radicals. Chem. Res. Toxicol. 22, 1747-1760.

(9)

Brookes, P., and Lawley, P. D. (1961) 766. The alkylation of guanosine and guanylic acid. J. Chem. Soc. (Resumed), 3923-3928.

(10)

Boiteux, S., Belleney, J., Roques, B. P., and Laval, J. (1984) Two rotameric forms of open ring 7-methylguanine are present in alkylated polnucleotides. Nucleic Acids Res. 12, 5429-5439.

(11)

O'Connor, T. R., Boiteux, S., and Laval, J. (1988) Ring-opened 7-methylguanine residues in DNA are a block to in vitro DNA synthesis. Nucleic Acids Res. 16, 5879-5894.

(12)

Lawley, P. D., and Brookes, P. (1961) Acidic Dissociation of 7 : 9-Dialkylguanines and its Possible Relation to Mutagenic Properties of Alkylating Agents. Nature 192, 10811082.

(13)

Chetsanga, C. J., and Lindahl, T. (1979) Release of 7-methylguanine residues whose imidazole rings have been opened from damaged DNA by a DNA glycosylase from Escherichia coli. Nucleic Acids Res. 6, 3673-3684.

(14)

Loeb, L. A., and Preston, B. D. (1986) Mutagenesis by apurinic/apyrimidinic sites. Annu. Rev. Genet. 20, 201-230.

(15)

Sowers, L. C., Sedwick, W. D., and Shaw, B. R. (1989) Hydrolysis of N3-methyl-2'deoxycytidine: model compound for reactivity of protonated cytosine residues in DNA. Mutat. Res. 215, 131-138.

28 ACS Paragon Plus Environment

Page 29 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(16)

Lawley, P. D., and Brookes, P. (1963) Further studies on the alkylation of nucleic acids and their constituent nucleotides. Biochem. J 89, 127-138.

(17)

Lawley, P. D., and Warren, W. (1976) Removal of minor methylation products 7methyladenine and 3-methylguanine from DNA of Escherichia coli treated with dimethyl sulphate. Chem. Biol. Interact. 12, 211-220.

(18)

Boiteux, S., Huisman, O., and Laval, J. (1984) 3-Methyladenine residues in DNA induce the SOS function sfiA in Escherichia coli. EMBO J. 3, 2569-2573.

(19)

Singer, B. (1972) Reaction of guanosine with ethylating agents. Biochemistry 11, 39393947.

(20)

Nooner, T., Dutta, S., and Gates, K. S. (2004) Chemical properties of the leinamycinguanine adduct in DNA. Chem. Res. Toxicol. 17, 942-949.

(21)

Guy, A., Molko, D., Wagrez, L., and Téoule, R. (1986) Chemical Synthesis of Oligonucleotides Containing N6-Methyladenine Residues in the GATC Site. Helv. Chim. Acta 69, 1034-1040.

(22)

Moschel, R. C., Hudgins, W. R., and Dipple, A. (1984) Substituent-induced effects on the stability of benzylated guanosines: model systems for the factors influencing the stability of carcinogen-modified nucleic acids. J. Org. Chem. 49, 363-372.

(23)

Müller, N., and Eisenbrand, G. (1985) The influence of N7 substituents on the stability of N7-alkylated guanosines. Chem.-Biol. Interact. 53, 173-181.

(24)

Hendler, S. S., Fuerer, E., and Srinivasan, P. R. (1970) Synthesis and chemical properties of monomers and polymers containing 7-methylguanine and an investigation of their

29 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 30 of 84

substrate or template properties for bacterial deoxyribonucleic acid or ribonucleic acid polymerases. Biochemistry 9, 4141-4153. (25)

D'Andrea, A. D., and Haseltine, W. A. (1978) Modification of DNA by aflatoxin B1 creates alkali-labile lesions in DNA at positions of guanine and adenine. Proc. Natl. Acad. Sci. U. S. A. 75, 4120-4124.

(26)

Coles, B. F., Welch, A. M., Hertzog, P. J., Lindsay, S. J., and Garner, R. C. (1980) Biological and chemical studies on 8,9-dihydroxy-8,9-dihydro-aflatoxin B1 and some of its esters. Carcinogenesis 1, 79-90.

(27)

Johnston, D. S., and Stone, M. P. (2000) Replication of a site-specific trans-8,9-dihydro8-(N7-guanyl)-9-hydroxyaflatoxin B(1) adduct by the exonuclease deficient klenow fragment of DNA polymerase I. Chem. Res. Toxicol. 13, 1158-1164.

(28)

Craddock, V. M., and Henderson, A. R. (1986) Effect of N-nitrosamines carcinogenic for oesophagus on O6-alkyl-guanine-DNA-methyl transferase in rat oesophagus and liver. J. Cancer Res. Clin. Oncol. 111, 229-236.

(29)

Margison, G. P., and O'Connor, P. J. (1979) Nucleic acid modification by N-nitroso compounds. , In Chemical carcinogens and DNA (P. L. Grover, Ed.) pp 11-159, CRC Press, Boca Raton, FL

(30)

Lawley, P. D. (1976) Carcinogenesis by alkylating agents, In Chemical Carcinogens ((ed.), S. C. E., Ed.) pp 83-224, American Chemical Society Monograph, 173, Washington, DC.

(31)

Miller, E. C., and Miller, J. A. (1981) Searches for ultimate chemical carcinogens and their reactions with cellular macromolecules. Cancer 47, 2327-2345. 30 ACS Paragon Plus Environment

Page 31 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(32)

Singer, B., and Kusmierek, J. T. (1982) Chemical mutagenesis. Annu. Rev. Biochem 51, 655-693.

(33)

Hsiung, H., and Lown, J. W. (1976) Effects of alkylation by dimethyl sulfate, nitrogen mustard, and mitomycin C on DNA structure as studied by the ethidium binding assay. Can. J. Biochem. 54, 1047-1054.

(34)

McGhee, J. D., and Felsenfeld, G. (1979) Reaction of nucleosome DNA with dimethyl sulfate. Proc. Natl. Acad. Sci. U. S. A. 76, 2133-2137.

(35)

Kyrtopoulos, S. A. (1998) DNA adducts in humans after exposure to methylating agents. Mutat. Res. 405, 135-143.

(36)

Kadlubar, F. F., Beranek, D. T., Weis, C. C., Evans, F. E., Cox, R., and Irving, C. C. (1984) Characterization of the purine ring-opened 7-methylguanine and its persistence in rat bladder epithelial DNA after treatment with the carcinogen N-methylnitrosourea. Carcinogenesis 5, 587-592.

(37)

Beranek, D. T., Weis, C. C., Evans, F. E., Chetsanga, C. J., and Kadlubar, F. F. (1983) Identification of N5-methyl-N5-formyl-2,5,6-triamino-4-hydroxypyrimidine as a major adduct in rat liver DNA after treatment with the carcinogens, N,N-dimethylnitrosamine or 1,2-dimethylhydrazine. Biochem. Biophys. Res. Commun. 110, 625-631.

(38)

Maccubbin, A. E., Caballes, L., Riordan, J. M., Huang, D. H., and Gurtoo, H. L. (1991) A cyclophosphamide/DNA phosphoester adduct formed in vitro and in vivo. Cancer Res. 51, 886-892.

31 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(39)

Page 32 of 84

Johnson, L. A., Malayappan, B., Tretyakova, N., Campbell, C., MacMillan, M. L., Wagner, J. E., and Jacobson, P. A. (2012) Formation of cyclophosphamide specific DNA adducts in hematological diseases. Pediatr. Blood Cancer 58, 708-714.

(40)

Moore, H. W. (1977) Bioactivation as a model for drug design bioreductive alkylation. Science 197, 527-532.

(41)

Tomasz, M., Lipman, R., Lee, M. S., Verdine, G. L., and Nakanishi, K. (1987) Reaction of acid-activated mitomycin C with calf thymus DNA and model guanines: elucidation of the base-catalyzed degradation of N7-alkylguanine nucleosides. Biochemistry 26, 20102027.

(42)

Hemminki, K. (1984) Reactions of ethyleneimine with guanosine and deoxyguanosine. Chem. Biol. Interact. 48, 249-260.

(43)

Hansen, M. R., and Hurley, L. H. (1996) Pluramycins. Old Drugs Having Modern Friends in Structural Biology. Acc. Chem. Res. 29, 249-258.

(44)

Coleman, R. S., Burk, C. H., Navarro, A., Brueggemeier, R. W., and Diaz-Cruz, E. S. (2002) Role of the azinomycin naphthoate and central amide in sequence-dependent DNA alkylation and cytotoxicity of epoxide-bearing substructures. Org. Lett. 4, 35453548.

(45)

Hartley, J. A., Hazrati, A., Kelland, L. R., Khanim, R., Shipman, M., Suzenet, F., and Walker, L. F. (2000) A Synthetic Azinomycin Analogue with Demonstrated DNA CrossLinking Activity: Insights into the Mechanism of Action of this Class of Antitumor Agent. Angew. Chem. Int. Ed. 39, 3467-3470.

32 ACS Paragon Plus Environment

Page 33 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(46)

Kim, M. S., and Guengerich, F. P. (1998) Polymerase blockage and misincorporation of dNTPs opposite the ethylene dibromide-derived DNA adducts S-[2-(N7guanyl)ethyl]glutathione, S-[2-(N2-guanyl)ethyl]glutathione, and S-[2-(O6guanyl)ethyl]glutathione. Chem. Res. Toxicol. 11, 311-316.

(47)

Valadez, J. G., and Guengerich, F. P. (2004) S-(2-chloroethyl)glutathione-generated p53 mutation spectra are influenced by differential repair rates more than sites of initial dna damage. J. Biol. Chem. 279, 13435-13446.

(48)

Evans, M. D., Dizdaroglu, M., and Cooke, M. S. (2004) Oxidative DNA damage and disease: induction, repair and significance. Mutat. Res. 567, 1-61.

(49)

Greenberg, M. M. (2012) The formamidopyrimidines: purine lesions formed in competition with 8-oxopurines from oxidative stress. Acc. Chem. Res. 45, 588-597.

(50)

Dizdaroglu, M., Kirkali, G., and Jaruga, P. (2008) Formamidopyrimidines in DNA: mechanisms of formation, repair, and biological effects. Free Radic. Biol. Med. 45, 16101621.

(51)

Hsu, I. C., Metcalf, R. A., Sun, T., Welsh, J. A., Wang, N. J., and Harris, C. C. (1991) Mutational hot spot in the p53 gene in human hepatocellular carcinomas. Nature 350, 427-428.

(52)

Bressac, B., Kew, M., Wands, J., and Ozturk, M. (1991) Selective G to T mutations of p53 gene in hepatocellular carcinoma from southern Africa. Nature 350, 429-431.

(53)

Boysen, G., Pachkowski, B. F., Nakamura, J., and Swenberg, J. A. (2009) The formation and biological significance of N7-guanine adducts. Mutat. Res. 678, 76-94.

33 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(54)

Page 34 of 84

Tudek, B. (2003) Imidazole ring-opened DNA purines and their biological significance. J. Biochem. Mol. Biol. 36, 12-19.

(55)

Rydberg, B., and Lindahl, T. (1982) Nonenzymatic methylation of DNA by the intracellular methyl group donor S-adenosyl-L-methionine is a potentially mutagenic reaction. EMBO J. 1, 211-216.

(56)

Humphreys, W. G., and Guengerich, F. P. (1991) Structure of formamidopyrimidine adducts as determined by NMR using specifically 15N-labeled guanosine. Chem. Res. Toxicol. 4, 632-636.

(57)

Christov, P. P., Brown, K. L., Kozekov, I. D., Stone, M. P., Harris, T. M., and Rizzo, C. J. (2008) Site-specific synthesis and characterization of oligonucleotides containing an N6-(2-deoxy-D-erythro-pentofuranosyl)-2,6-diamino-3,4-dihydro-4-oxo-5-N-methylfor mamidopyrimidine lesion, the ring-opened product from N7-methylation of deoxyguanosine. Chem. Res. Toxicol. 21, 2324-2333.

(58)

van Delft, J. H., van Weert, E. J., Schellekens, M. M., Claassen, E., and Baan, R. A. (1991) The isolation of monoclonal antibodies selected for the detection of imidazole ring-opened N7-ethylguanine in purified DNA and in cells in situ. Crossreaction with methyl, 2-hydroxyethyl and sulphur mustard adducts. Carcinogenesis 12, 1041-1049.

(59)

Farmer, P. B., Foster, A. B., Jarman, M., and Tisdale, M. J. (1973) The alkylation of 2'deoxyguanosine and of thymidine with diazoalkanes. Some observations on o-alkylation. The Biochemical journal 135, 203-213.

(60)

Segerback, D. (1983) Alkylation of DNA and hemoglobin in the mouse following exposure to ethene and ethene oxide. Chem. Biol. Interact. 45, 139-151.

34 ACS Paragon Plus Environment

Page 35 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(61)

Filser, J. G., Denk, B., Tornqvist, M., Kessler, W., and Ehrenberg, L. (1992) Pharmacokinetics of ethylene in man; body burden with ethylene oxide and hydroxyethylation of hemoglobin due to endogenous and environmental ethylene. Arch. Toxicol. 66, 157-163.

(62)

Walker, V. E., Fennell, T. R., Boucheron, J. A., Fedtke, N., Ciroussel, F., and Swenberg, J. A. (1990) Macromolecular adducts of ethylene oxide: a literature review and a timecourse study on the formation of 7-(2-hydroxyethyl)guanine following exposures of rats by inhalation. Mutat. Res. 233, 151-164.

(63)

Roe, R., Paul, J. S., and Montgomery, P. O. B. (1973) Synthesis and PMR spectra of 7hydroxyalkylguanosinium acetates. J. Heterocycl. Chem. 10, 849-857.

(64)

Lee, F. I., and Harry, D. S. (1974) Angiosarcoma of the liver in a vinyl-chloride worker. Lancet 303, 1316–1318.

(65)

Marsteller, H. J., Lelbach, W. K., Muler, R., and Gedigk, P. (1975) Unusual splenomegalic liver disease as evidenced by peritoneoscopy and guided liver biopsy among polyvinyl chloride production workers. Ann. N. Y. Acad. Sci. 246, 95-134.

(66)

Nicholson, W. J., Hammond, E. C., Seidman, H., and Selikoff, I. J. (1975) Mortality experience of a cohort of vinyl chloride-polyvinyl chloride workers. Ann. N. Y. Acad. Sci. 246, 225-230.

(67)

Creech, J. L., Jr., and Johnson, M. N. (1974) Angiosarcoma of liver in the manufacture of polyvinyl chloride. J. Occup. Med. 16, 150-151.

35 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(68)

Page 36 of 84

Guengerich, F. P., Crawford, W. M., and Watanabe, P. G. (1979) Activation of vinyl chloride to covalently bound metabolites: roles of 2-chloroethylene oxide and 2chloroacetaldehyde. Biochemistry 18, 5177-5182.

(69)

Christov, P. P., Kozekov, I. D., Rizzo, C. J., and Harris, T. M. (2008) The formamidopyrimidine derivative of 7-(2-oxoethyl)-2'-deoxyguanosine. Chem. Res. Toxicol. 21, 1777-1786.

(70)

Hata, Y., Watanabe, M., Tonda, K., and Hirata, M. (1987) Aziridine biotransformation by microsomes and lethality to hepatocytes isolated from rat. Chem. Biol. Interact. 63, 171184.

(71)

Verschaeve, L., and Kirsch-Volders, M. (1990) Mutagenicity of ethyleneimine. Mutat. Res. 238, 39-55.

(72)

Bosies, E., Endele, R., and Pahlke, W. (1989) N-Substituted aziridine-2-carboxylic acid derivatives, processes for the preparation thereof and pharmaceutical compositions containing them, In United States Patent Boehringer Mannheim, GmbH, USA.

(73)

Chabner, B. A., and Roberts, T. G., Jr. (2005) Timeline: Chemotherapy and the war on cancer. Nat. Rev. Cancer 5, 65-72.

(74)

Emadi, A., Jones, R. J., and Brodsky, R. A. (2009) Cyclophosphamide and cancer: golden anniversary. Nat. Rev. Clin. Oncol. 6, 638-647.

(75)

Loeber, R. L., Michaelson-Richie, E. D., Codreanu, S. G., Liebler, D. C., Campbell, C. R., and Tretyakova, N. Y. (2009) Proteomic analysis of DNA-protein cross-linking by antitumor nitrogen mustards. Chem. Res. Toxicol. 22, 1151-1162.

36 ACS Paragon Plus Environment

Page 37 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(76)

Yule, S. M., Foreman, N. K., Mitchell, C., Gouldon, N., May, P., and McDowell, H. P. (1997) High-dose cyclophosphamide for poor-prognosis and recurrent pediatric brain tumors: a dose-escalation study. J. Clin. Oncol. 15, 3258-3265.

(77)

Kohn, K. W., Spears, C. L., and Doty, P. (1966) Inter-strand crosslinking of DNA by nitrogen mustard. J. Mol. Biol. 19, 266-288.

(78)

Chetsanga, C. J., Polidori, G., and Mainwaring, M. (1982) Analysis and excision of ringopened phosphoramide mustard-deoxyguanine adducts in DNA. Cancer Res. 42, 26162621.

(79)

Gruppi, F., Hejazi, L., Christov, P. P., Krishnamachari, S., Turesky, R. J., and Rizzo, C. J. (2015) Characterization of nitrogen mustard formamidopyrimidine adduct formation of bis(2-chloroethyl)ethylamine with calf thymus DNA and a human mammary cancer cell line. Chem. Res. Toxicol. 28, 1850-1860.

(80)

Christov, P. P., Son, K. J., and Rizzo, C. J. (2014) Synthesis and characterization of oligonucleotides containing a nitrogen mustard formamidopyrimidine monoadduct of deoxyguanosine. Chem. Res. Toxicol. 27, 1610-1618.

(81)

Busby, W. F. J., and Wogan, G. N. (1984) Chemical Carcinogens, (Searle, C., Ed.) pp 945-1136, American Chemical Society, Washington, DC.

(82)

Brown, K. L., Deng, J. Z., Iyer, R. S., Iyer, L. G., Voehler, M. W., Stone, M. P., Harris, C. M., and Harris, T. M. (2006) Unraveling the Aflatoxin−FAPY Conundrum:  Structural Basis for Differential Replicative Processing of Isomeric Forms of the Formamidopyrimidine-Type DNA Adduct of Aflatoxin B1. J. Am. Chem. Soc. 128, 15188-15199.

37 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(83)

Page 38 of 84

Croy, R. G., and Wogan, G. N. (2008) Temporal Patterns of Covalent DNA Adducts in Rat Liver after Single and Multiple Doses of Aflatoxin B1. Cancer Res. 41, 197-203.

(84)

Essigmann, J. M., Croy, R. G., Bennett, R. A., and Wogan, G. N. (1982) Metabolic Activation of Aflatoxin B1: Patterns of DNA Adduct Formation, Removal, and Excretion in Relation to Carcinogenesis. Drug Metab. Rev. 13, 581-602.

(85)

Foster, P. L., Eisenstadt, E., and Miller, J. H. (1983) Base substitution mutations induced by metabolically activated aflatoxin B1. Proc. Natl. Acad. Sci. U. S. A. 80, 2695-2698.

(86)

McMahon, G., Davis, E. F., Huber, L. J., Kim, Y., and Wogan, G. N. (1990) Characterization of c-Ki-ras and N-ras oncogenes in aflatoxin B1-induced rat liver tumors. Proc. Natl. Acad. Sci. U. S. A 87, 1104-1108.

(87)

Chang, Y. J., Mathews, C., Mangold, K., Marien, K., Hendricks, J., and Bailey, G. (1991) Analysis of ras gene mutations in rainbow trout liver tumors initiated by aflatoxin B1. Mol. Carcinog. 4, 112-119.

(88)

Trottier, Y., Waithe, W. I., and Anderson, A. (1992) Kinds of mutations induced by aflatoxin B1 in a shuttle vector replicating in human cells transiently expressing cytochrome P4501A2 cDNA. Mol. Carcinog. 6, 140-147.

(89)

Lin, J. K., Miller, J. A., and Miller, E. C. (1977) 2,3-Dihydro-2-(guan-7-yl)-3-hydroxyaflatoxin B1, a major acid hydrolysis product of aflatoxin B1-DNA or -ribosomal RNA adducts formed in hepatic microsome-mediated reactions and in rat liver in vivo. Cancer Res. 37, 4430-4438.

38 ACS Paragon Plus Environment

Page 39 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(90)

Hertzog, P. J., Smith, J. R. L., and Garner, R. C. (1982) Characterisation of the imidazole ring-opened forms of trans-8,9-di-hydro-8-(7-guanyl)9-hydroxy aflatoxin B1. Carcinogenesis 3, 723-725.

(91)

Bailey, E. A., Iyer, R. S., Stone, M. P., Harris, T. M., and Essigmann, J. M. (1996) Mutational properties of the primary aflatoxin B1-DNA adduct. Proc. Natl. Acad. Sci. U. S. A. 93, 1535-1539.

(92)

Lawrence, C. W., Borden, A., Banerjee, S. K., and LeClerc, J. E. (1990) Mutation frequency and spectrum resulting from a single abasic site in a single-stranded vector. Nucleic Acids Res. 18, 2153-2157.

(93)

Leadon, S. A., and Snowden, M. M. (1988) Differential repair of DNA damage in the human metallothionein gene family. Mol. Cell. Biol. 8, 5331-5338.

(94)

Kaden, D. A., Call, K. M., Leong, P. M., Komives, E. A., and Thilly, W. G. (1987) Killing and mutation of human lymphoblast cells by aflatoxin B1: evidence for an inducible repair response. Cancer Res. 47, 1993-2001.

(95)

Ball, R. W., Wilson, D. W., and Coulombe, R. A., Jr. (1990) Comparative formation and removal of aflatoxin B1-DNA adducts in cultured mammalian tracheal epithelium. Cancer Res. 50, 4918-4922.

(96)

Smela, M. E., Hamm, M. L., Henderson, P. T., Harris, C. M., Harris, T. M., and Essigmann, J. M. (2002) The aflatoxin B(1) formamidopyrimidine adduct plays a major role in causing the types of mutations observed in human hepatocellular carcinoma. Proc. Natl. Acad. Sci. U. S. A. 99, 6655-6660.

39 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(97)

Page 40 of 84

Gopalakrishnan, S., and Patel, D. J. (1993) Formation and structural features of a sterigmatocystin-formamidopyrimidine adduct at the DNA duplex level. J. Am. Chem. Soc. 115, 9321-9322.

(98)

Baertschi, S. W., Raney, K. D., Stone, M. P., and Harris, T. M. (1988) Preparation of the 8,9-epoxide of the mycotoxin aflatoxin B1: the ultimate carcinogenic species. J. Am. Chem. Soc. 110, 7929-7931.

(99)

Gopalakrishnan, S., Stone, M. P., and Harris, T. M. (1989) Preparation and characterization of an aflatoxin B1 adduct with the oligodeoxynucleotide d(ATCGAT)2. J. Am. Chem. Soc. 111, 7232-7239.

(100) Gopalakrishnan, S., Harris, T. M., and Stone, M. P. (1990) Intercalation of aflatoxin B1 in two oligodeoxynucleotide adducts: comparative 1H NMR analysis of d(ATCAFBGAT).d(ATCGAT) and d(ATAFBGCAT)2. Biochemistry 29, 10438-10448. (101) Szybalski, W., and Iyer, V. N. (1967) Mechanisms of Action In Antibiotics I (Gottleib, D., and Shaw, P. D., Eds.) p 230, Springer-Verlag, New York. (102) Berger, M., and Cadet, J. (1985) Isolation and Characterization of the Radiation-Induced Degradation Products of 2′-Deoxyguanosine in Oxygen-Free Aqueous Solutions, In Z. Naturforsch. B p 1519. (103) Haraguchi, K., and Greenberg, M. M. (2001) Synthesis of Oligonucleotides Containing Fapy·dG (N6- (2-Deoxy-α,β-d-erythro-pentofuranosyl)-2,6- diamino-4-hydroxy-5formamidopyrimidine). J. Am. Chem. Soc. 123, 8636-8637.

40 ACS Paragon Plus Environment

Page 41 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(104) Tudek, B., Graziewicz, M., Kazanova, O., Zastawny, T. H., Obtulowicz, T., and J., L. (1999) Mutagenic specificity of imidazole ring-opened 7-metbylpurines in M13mp18 phage DNA. Acta. Biochim. Polo. 46, 785-799. (105) Ober, M., Linne, U., Gierlich, J., and Carell, T. (2003) The two main DNA lesions 8Oxo-7,8-dihydroguanine and 2,6-diamino-5-formamido-4-hydroxypyrimidine exhibit strongly different pairing properties. Angew. Chem. Int. Ed. 42, 4947-4951. (106) Ober, M., Muller, H., Pieck, C., Gierlich, J., and Carell, T. (2005) Base pairing and replicative processing of the formamidopyrimidine-dG DNA lesion. J. Am. Chem. Soc. 127, 18143-18149. (107) Coste, F., Ober, M., Le Bihan, Y. V., Izquierdo, M. A., Hervouet, N., Mueller, H., Carell, T., and Castaing, B. (2008) Bacterial base excision repair enzyme Fpg recognizes bulky N7-substituted-FapydG lesion via unproductive binding mode. Chem. Biol. 15, 706-717. (108) Domínguez, B. M., and Cullis, P. M. (1999) 2-Azabicyclo[2.2.1]hept-5-en-3-one epoxide: A versatile intermediate for the synthesis of cyclopentyl carbocyclic 2-deoxy-, 3-deoxy- & ara- ribonucleoside analogues. Tetrahedron Lett. 40, 5783-5786. (109) Boiteux, S., and Laval, J. (1983) Imidazole open ring 7-methylguanine: an inhibitor of DNA synthesis. Biochem. Biophys. Res. Commun. 110, 552-558. (110) Asagoshi, K., Terato, H., Ohyama, Y., and Ide, H. (2002) Effects of a guanine-derived formamidopyrimidine lesion on DNA replication: translesion DNA synthesis, nucleotide insertion, and extension kinetics. J. Biol. Chem. 277, 14589-14597. (111) Tudek, B., Boiteux, S., and Laval, J. (1992) Biological properties of imidazole ringopened N7-methylguanine in M13mp18 phage DNA. Nucleic Acids Res. 20, 3079-3084. 41 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 42 of 84

(112) Christov, P. P., Yamanaka, K., Choi, J.-Y., Takata, K.-i., Wood, R. D., Guengerich, F. P., Lloyd, R. S., and Rizzo, C. J. (2012) Replication of the 2,6-Diamino-4-hydroxy-N5(methyl)-formamidopyrimidine (MeFapy-dGuo) Adduct by Eukaryotic DNA Polymerases. Chem. Res. Toxicol. 25, 1652-1661. (113) Grziewicz, M.-A., Zastawny, T. H., Oliński, R., and Tudek, B. (1999) SOS-dependent A→G transitions induced by hydroxyl radical generating system hypoxanthine/xanthine oxidase/Fe3+/EDTA are accompanied by the increase of Fapy-adenine content in M13 mp18 phage DNA. Mutat. Res./DNA Repair 434, 41-52. (114) Smela, M. E., Currier, S. S., Bailey, E. A., and Essigmann, J. M. (2001) The chemistry and biology of aflatoxin B(1): from mutational spectrometry to carcinogenesis. Carcinogenesis 22, 535-545. (115) Boiteux, S., Gajewski, E., Laval, J., and Dizdaroglu, M. (1992) Substrate specificity of the Escherichia coli Fpg protein (formamidopyrimidine-DNA glycosylase): excision of purine lesions in DNA produced by ionizing radiation or photosensitization. Biochemistry 31, 106-110. (116) Breimer, L. H. (1984) Enzymatic excision from γ-irradiated polydeoxyribonucleotids of adenine residues whose imidazole rings have been rypture. Nucleic Acids Res. 12, 63596367. (117) Boiteux, S., O'Connor, T. R., and Laval, J. (1987) Formamidopyrimidine-DNA glycosylase of Escherichia coli: cloning and sequencing of the fpg structural gene and overproduction of the protein. Embo. J 6, 3177-3183.

42 ACS Paragon Plus Environment

Page 43 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(118) Laval, J., Boiteux, S., and O'Connor, T. R. (1990) Physiological properties and repair of apurinic/apyrimidinic sites and imidazole ring-opened guanines in DNA. Mutat. Res. 233, 73-79. (119) Boiteux, S., O'Connor, T. R., Lederer, F., Gouyette, A., and Laval, J. (1990) Homogeneous Escherichia coli FPG protein. A DNA glycosylase which excises imidazole ring-opened purines and nicks DNA at apurinic/apyrimidinic sites. J. Biol. Chem. 265, 3916-3922. (120) Tudek, B., Van Zeeland, A. A., Kusmierek, J. T., and Laval, J. (1998) Activity of Escherichia coli DNA-glycosylases on DNA damaged by methylating and ethylating agents and influence of 3-substituted adenine derivatives. Mutat. Res. 407, 169-176. (121) van der Kemp, P. A., Thomas, D., Barbey, R., de Oliveira, R., and Boiteux, S. (1996) Cloning and expression in Escherichia coli of the OGG1 gene of Saccharomyces cerevisiae, which codes for a DNA glycosylase that excises 7,8-dihydro-8-oxoguanine and 2,6-diamino-4-hydroxy-5-N-methylformamidopyrimidine. Proc. Natl. Acad. Sci. U. S. A. 93, 5197-5202. (122) Karahalil, B., Girard, P. M., Boiteux, S., and Dizdaroglu, M. (1998) Substrate specificity of the Ogg1 protein of Saccharomyces cerevisiae: excision of guanine lesions produced in DNA by ionizing radiation- or hydrogen peroxide/metal ion-generated free radicals. Nucleic Acids Res. 26, 1228-1233. (123) Hazra, T. K., Kow, Y. W., Hatahet, Z., Imhoff, B., Boldogh, I., Mokkapati, S. K., Mitra, S., and Izumi, T. (2002) Identification and characterization of a novel human DNA glycosylase for repair of cytosine-derived lesions. J. Biol. Chem. 277, 30417-30420.

43 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 44 of 84

(124) Hazra, T. K., Izumi, T., Boldogh, I., Imhoff, B., Kow, Y. W., Jaruga, P., Dizdaroglu, M., and Mitra, S. (2002) Identification and characterization of a human DNA glycosylase for repair of modified bases in oxidatively damaged DNA. Proc. Natl. Acad. Sci. U. S. A. 99, 3523-3528. (125) Bandaru, V., Sunkara, S., Wallace, S. S., and Bond, J. P. (2002) A novel human DNA glycosylase that removes oxidative DNA damage and is homologous to Escherichia coli endonuclease VIII. DNA Repair (Amst) 1, 517-529. (126) Takao, M., Kanno, S., Kobayashi, K., Zhang, Q. M., Yonei, S., van der Horst, G. T., and Yasui, A. (2002) A back-up glycosylase in Nth1 knock-out mice is a functional Nei (endonuclease VIII) homologue. J. Biol. Chem. 277, 42205-42213. (127) Izumi, T., Wiederhold, L. R., Roy, G., Roy, R., Jaiswal, A., Bhakat, K. K., Mitra, S., and Hazra, T. K. (2003) Mammalian DNA base excision repair proteins: their interactions and role in repair of oxidative DNA damage. Toxicology 193, 43-65. (128) Doetsch, P. W., and Cunningham, R. P. (1990) The enzymology of apurinic/apyrimidinic endonucleases. Mutat. Res./DNA Repair 236, 173-201. (129) Ramotar, D. The apurinic-apyrimidinic endonuclease IV family of DNA repair enzymes. Biochem. Cell Biol. 75, 327–336. (130) Ljungquist, S., Lindahl, T., and Howard-Flanders, P. (1976) Methyl methane sulfonatesensitive mutant of Escherichia coli deficient in an endonuclease specific for apurinic sites in deoxyribonucleic acid. J. Bacteriol. 126, 646-653. (131) Kerins, S. M., Collins, R., and McCarthy, T. V. (2003) Characterization of an Endonuclease IV 3′-5′ Exonuclease Activity. J. Biol. Chem. 278, 3048-3054. 44 ACS Paragon Plus Environment

Page 45 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

(132) Golan, G., Ishchenko, A. A., Khassenov, B., Shoham, G., and Saparbaev, M. K. (2010) Coupling of the nucleotide incision and 3′ → 5′ exonuclease activities in Escherichia coli endonuclease IV: Structural and genetic evidences. Mutat. Res. Fund. Mol. Mech. Mut. 685, 70-79. (133) Ide, H., Tedzuka, K., Shimzu, H., Kimura, Y., Purmal, A. A., Wallace, S. S., and Kow, Y. W. (1994).alpha.-Deoxyadenosine, a Major Anoxic Radiolysis Product of Adenine in DNA, Is a Substrate for Escherichia coli Endonuclease IV. Biochemistry 33, 7842-7847. (134) Aramini, J. M., Cleaver, S. H., Pon, R. T., Cunningham, R. P., and Germann, M. W. (2004) Solution Structure of a DNA Duplex Containing an α-Anomeric Adenosine: Insights into Substrate Recognition by Endonuclease IV. J. Mol. Biol. 338, 77-91. (135) Gros, L., Ishchenko, A. A., Ide, H., Elder, R. H., and Saparbaev, M. K. (2004) The major human AP endonuclease (Ape1) is involved in the nucleotide incision repair pathway. Nucleic Acids Res. 32, 73-81. (136) Daviet, S., Couvé-Privat, S., Gros, L., Shinozuka, K., Ide, H., Saparbaev, M., and Ishchenko, A. A. (2007) Major oxidative products of cytosine are substrates for the nucleotide incision repair pathway. DNA Repair 6, 8-18. (137) Patro, J. N., Haraguchi, K., Delaney, M. O., and Greenberg, M. M. (2004) Probing the Configurations of Formamidopyrimidine Lesions Fapy·dA and Fapy·dG in DNA Using Endonuclease IV. Biochemistry 43, 13397-13403. (138) Asagoshi, K., Yamada, T., Terato, H., Ohyama, Y., Monden, Y., Arai, T., Nishimura, S., Aburatani, H., Lindahl, T., and Ide, H. (2000) Distinct Repair Activities of Human 7,8Dihydro-8-oxoguanine DNA Glycosylase and Formamidopyrimidine DNA Glycosylase

45 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 46 of 84

for Formamidopyrimidine and 7,8-Dihydro-8-oxoguanine. J. Biol. Chem. 275, 49564964. (139) Christov, P. P., Banerjee, S., Stone, M. P., and Rizzo, C. J. (2010) Selective Incision of the α-N(5)-Methyl-Formamidopyrimidine Anomer by Escherichia coli Endonuclease IV. J. Nucleic Acids. 2010, 850234. (140) Alekseyev, Y. O., Hamm, M. L., and Essigmann, J. M. (2004) Aflatoxin B1 formamidopyrimidine adducts are preferentially repaired by the nucleotide excision repair pathway in vivo. Carcinogenesis 25, 1045-1051.

46 ACS Paragon Plus Environment

Page 47 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Biographies:

Dr. Suresh S. Pujari received his M.Sc. from Karnatak University, Dharwad, India and worked as a project assistant at National Chemical Laboratory, Pune, India. He pursued his Ph.D. from the University of Osnabrueck, Germany (Dr. Frank Seela) in Chemistry. After Ph.D, he worked as a postdoctoral fellow at Clemson University, Clemson, USA. Currently, he is working as a postdoctoral associate in Dr. Tretyakova's group at the Department of Medicinal Chemistry, Masonic Cancer Center and the College of Pharmacy, University of Minnesota, USA.

Dr. Natalia Tretyakova received her M.S. degree from Moscow State University in Russia and her Ph.D. from the University of North Carolina-Chapel Hill, where she worked with James A. Swenberg. She did her postdoctoral work with Steven R. Tannenbaum at the Massachusetts Institute of Technology. She is a Professor of Medicinal Chemistry at the Masonic Cancer Center and the College of Pharmacy, University of Minnesota.

47 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 48 of 84

Figure legends:

Figure 1. 500 MHz NMR spectra of N5-methyl-N5-formyl-2,5,6-triamino-4-hydroxypyrimidine. Spectra were obtained in DMSO-d6/D2O (1:1) (a), in MeOH-d4 (b); and in DMSO-d6 (c). Kadlubar, F. F., Beranek, D. T., Weis, C. C., Evans, F. E., Cox, R., and Irving, C. C., Characterization of the purine ring-opened 7-methylguanine and its persistence in rat bladder epithelial DNA after treatment with the carcinogen N-methylnitrosourea, Carcinogenesis, 1984, Vol. 5, Issue 5, pages 587-592, by permission of Oxford University Press.36 Figure 2. Rotational isomers of Methyl-FAPy-Gua adducts identified by Beranek et al.36, 37 Figure 3. Proton NMR spectra showing formamido signals with methylene protons of MethylFapy isomers. Spectra were taken in DMSO-d6. Boiteux, S., Belleney, J., Roques, B. P., and Laval, J., Two rotameric forms of open ring 7-methylguanine are present in alkylated polnucleotides, Nucleic Acids Res., 1984, Vol. 12, Issue 13, pages 5429-5439, by permission of Oxford University Press.10 Figure 4. UV absorption spectra of (A) 7-Et-Guo in water (—), 0.1 N HCl (--) and 0.1N KOH (….). Spectrum shown for 0.1N KOH is for Et-FAPy-G. (B) Ethyl-FAPy-Guo in 0.1 N KOH (….), 0.1N HCl (--).19 Figure 5. Structures of nitrogen mustards. Figure 6. Structures of aflatoxin B1 epoxide 26, its cationic N7-dG adduct 27, and AFB-FAPydG 28. Figure 7. Proposed structures of AFB-FAPy-dG isomers put forward by Miller and others.89 48 ACS Paragon Plus Environment

Page 49 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Figure 8. Structures of AFB-FAPy isomers proposed by Hertzog et. al.90 Figure 9. Structures of AFB-FAPy isomers elucidated by Harris et al.82 Figure 10. Structural isomers of MMC-FAPy-dG. Figure 11. HPLC analysis of ODN 5'-d(TT-Me-FAPy-dG)-TTC-3'. (A) Analysis of ODN synthesis with a short deprotection cycle. (B) Analysis of ODN synthesis with a long deprotection cycle. In both figures, peak 1 represents the formation of pyranose adduct, whereas other peaks are mixture of isomers.57 Figure 12. Structures of N5-R-FAPy lesions investigated in biological studies. Figure 13. Structures of N7-AFB1-G and AFB1-FAPy lesions investigated in biological studies.82 Figure 14. Rotamers of Bz-cFAPy-dG.107

49 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 50 of 84

Figure 1

50 ACS Paragon Plus Environment

Page 51 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Figure 2

OH N H2N

N

CH3 N CHO NH2

OH

CHO N CH3

N

NH2

N H2N

1

OH N H2N

2

CH3 N H O NH2

N 3

OH N H2N

CH3 N O H NH2

N 4

51 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 52 of 84

Figure 3

52 ACS Paragon Plus Environment

Page 53 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Figure 4

53 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 54 of 84

Figure 5

54 ACS Paragon Plus Environment

Page 55 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Figure 6

H3CO

H3CO

H3CO

O

O

O O

O

O

H

O

H HO O

O

26

O

O

O

N

N dR 27

O

O

O

H HO O

O N

HN H 2N

H

N

HN H 2N

H O

N

CHO NH dR

28

55 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 56 of 84

Figure 7

56 ACS Paragon Plus Environment

Page 57 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Figure 8

57 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 58 of 84

Figure 9

58 ACS Paragon Plus Environment

Page 59 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Figure 10

H2N

H2N

O

CH3

O

H2N CH3

H2N CH3

O

O

CH3

O

O

O

O H2NOCO MeO H O 1' N

HN H2 N

N

N

N

H2NOCO MeO H O

NH2

N

HN H2N HO

NH H

N

H2NOCO MeO H

NH2

O

O

H2NOCO MeO H

NH2

O

N

HN H2N

N

NH H

N OH

O

HN H2N

N

NH2 N

O

NH H

N O

O

1'-alpha+1'-beta isomers

O OH

47

N

HO

48

HO HO 49

50

59 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 60 of 84

Figure 11

60 ACS Paragon Plus Environment

Page 61 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Figure 12

61 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 62 of 84

Figure 13

62 ACS Paragon Plus Environment

Page 63 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Figure 14

63 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 64 of 84

Scheme legends:

Scheme 1. Mechanisms leading to the formation of FAPy adducts in DNA. Scheme 2. Formation of methyl-FAPy from dG. Scheme 3. Synthesis of Me-FAPy-dG from N2, 5'-OH -protected dG.57 Scheme 4. Synthetic scheme for Ethyl-FAPy-dG.58 Scheme 5. Synthesis of N7-(2-hydroxyethyl)-FAPy-dG.63 Scheme 6. Synthesis of 2-oxyethyl-FAPy-dG.69 Scheme 7. Synthesis of Ethylamine-FAPy-dG.42 Scheme 8. Synthesis of NM-FAPy-dG by Christov et. al.80 Scheme 9. Synthesis of AFB-FAPy-dG.82 Scheme 10. FAPy formation of sterigmatocystin 1 epoxide.97 Scheme 11. Synthesis of MMC-FAPy-dG.41 Scheme 12. Isomerization of N5-R-FAPy adducts to α, β anomers 51, 53 and pyranose derivatives 54 via an imine intermediate. Scheme 13. Direct treatment of DNA containing a single dG residue to introduce N5-R-FAPyresidue. Scheme 14. Synthesis of AFB-FAPy-dG oligonucleotide.82 Scheme 15. Direct methylation of M13 phage DNA to form Me-FAPy adducts.111

64 ACS Paragon Plus Environment

Page 65 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Scheme 16. Preparation of DNA containing radilabaled phosphoramide mustard-FAPy adducts.104 Scheme 17. Synthesis of Me-FAPy-dG phosphoramidite.57 Scheme 18. Preparation of NM-FAPy-dG phosphoramidite building block for solid phase synthesis.80 Scheme 19. Synthesis of Bz-cFAPy-dG phosphoramidite.105-107

65 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 66 of 84

Scheme 1

66 ACS Paragon Plus Environment

Page 67 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Scheme 2

O

H 2N HO

O N

HN

N

N

HN methylating agents

H 2N

CH3

N

N

HO O OH

OH

CH3 N

HN

CHO NH

OH H2N

N

HO

O OH

O

N

O OH

OH

OH H

O HN H 2N

CH3 N

N

CHO NH2

1

67 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 68 of 84

Scheme 3

68 ACS Paragon Plus Environment

Page 69 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Scheme 4

69 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 70 of 84

Scheme 5

70 ACS Paragon Plus Environment

Page 71 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Scheme 6

71 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 72 of 84

Scheme 7

72 ACS Paragon Plus Environment

Page 73 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Scheme 8

73 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 74 of 84

Scheme 9

74 ACS Paragon Plus Environment

Page 75 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Scheme 10

75 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 76 of 84

Scheme 11

76 ACS Paragon Plus Environment

Page 77 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Scheme 12

77 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 78 of 84

Scheme 13

78 ACS Paragon Plus Environment

Page 79 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Scheme 14

79 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 80 of 84

Scheme 15

80 ACS Paragon Plus Environment

Page 81 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Scheme 16

81 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 82 of 84

Scheme 17

82 ACS Paragon Plus Environment

Page 83 of 84

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Chemical Research in Toxicology

Scheme 18

OAc

Et

N

N 25 CsOAc 18-crown-6 2h, toulene

(H3C)2N N DMTO

N OH

O

O CHO

NH

N O OH 57

N

HN

TBAF (H3C)2N N THF, CH2Cl2 DMTO

OAc

Et

N

N

(H3C)3Si

OAc

Et

NH

N

N

N

CHO NC(CH2)2OP[N(i-Pr)2] (H3C)2N N DMTO 1H-tetrazole, CH2Cl2

N O

O OH 58

NC(H2C)2O

CHO

NH

O P

N(i-Pr)2

59

83 ACS Paragon Plus Environment

Chemical Research in Toxicology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 84 of 84

Scheme 19

84 ACS Paragon Plus Environment